You are on page 1of 16

Pathophysiology of Fetal

and Neonatal Kidneys

Farid Boubred and Umberto Simeoni

Abstract Contents
Fetal renal development is a complex phenom-
Kidney Development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
enon. The ureteric bud and metanephric mesen-
chyme interaction is essential for nephrogenesis Renal Physiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
The Fetus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
which results from the expression of specific Glomerular Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
genes, the fetal environment, or the interaction Tubular Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
of both factors. IUGR, maternal diabetes, mater- The Newborn . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
nal undernutrition, micronutrient deficiency, and Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
fetal or neonatal exposure to drugs (NSAIDS, The Immature Kidney . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
ACE-Is, glucocorticoids, aminoglycosides) or to Congenital Tubulopathies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Parenchymal Consequences of Urinary Tract
toxic (alcohol) are known to affect
Anomalies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
nephrogenesis. After a brief review of fetal and Pharmacological Interactions . . . . . . . . . . . . . . . . . . . . . . . . . 10
neonatal renal physiology, pathologic situations Long-Term Consequences of Nephron Number
including preterm birth, congenital anomalies of Reduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
the kidney and urinary tract, kidney diseases, References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
and kidney-pharmacology interactions will be
developed. Renal maldevelopment and expo-
sure to factors that can alter the fetal and neona- Salient Points
tal renal functions and structure increase the risk
of hypertension and chronic renal disease in • Fetal renal development starts with an interac-
adulthood. Long-term follow-up is thus required tion between the ureteric bud and metanephric
and early markers of nephron dosing and renal mesenchyme and an interaction between the
injury should be developed with the aim to expression of specific genes and the fetal
implement preventive strategies. environment.
• During intrauterine life, the placenta is respon-
sible for fetal homeostasis. Glomerular filtra-
F. Boubred tion rate, renal blood flow, and tubular
Division of Neonatology, La Conception Hospital, functions increase with renal growth and
Marseille, France nephrogenesis. The formation of the urine,
e-mail: farid.boubred@ap-hm.fr
the main component of amniotic fluid in the
U. Simeoni (*) third trimester, begins by 12 weeks of gesta-
Division of Pediatrics, CHUV & UNIL, Lausanne, Vaud,
tion. Urine production is essential for fetal
Switzerland
e-mail: Umberto.simeoni@chuv.ch well-being, the promotion of lung growth,

# Springer International Publishing AG 2018 1


G. Buonocore et al. (eds.), Neonatology,
https://doi.org/10.1007/978-3-319-18159-2_261-2
2 F. Boubred and U. Simeoni

and in hormonal production (1.25 OH vitD3 should be developed with the aim of
and erythropoietin). Urine flow increases ten- implementing preventive strategies.
fold from 6 ml/h at 20 weeks to 60 ml/h at 40
weeks of gestation. At near-term, the fetal kid-
ney shows sufficient glomerular and tubular Kidney Development
development to allow for adaptation to extra-
uterine life. The definitive kidney, the metanephros, is formed
• Serum creatinine is used to assess glomerular by two processes: nephrogenesis (formation of
filtration rate in neonates. glomerulus and tubules) and branching morpho-
• At birth, the final nephron number varies from genesis (formation of collecting ducts, calyces,
500,000 to 1 million per kidney. Such variation pelvis, and ureter). The metanephric kidney
in nephron number is due to genetic factors, the takes place after the formation and involution of
fetal environment, or a combination of both. two embryonic kidneys, the pronephros (non-
Intrauterine growth restriction, maternal diabe- functional organ) and mesonephros (first func-
tes, maternal undernutrition, micronutrient tional kidney), that in turn evolves into the
deficiency, and fetal or neonatal exposure to ureteric bud (UB). The metanephros appears in
drugs (NSAIDS, ACE-Is, glucocorticoids, the fifth gestational week and develops from the
aminoglycosides) or toxins (alcohol) affect specific interaction of the epithelial UB and the
nephrogenesis. undifferentiated metanephric mesenchyme (MM).
• In preterm infants, nephrogenesis is incom- This interaction is crucial for the differentiation of
plete at birth and may be altered in preterm mesenchyme and the induction of UB branching
infants. division (Fig. 1). The UB undergoes branching
• Congenital tubular diseases are rare. Bartter morphogenesis and invades the MM in response
syndrome (characterized by salt wasting, to signals elaborated by the mesenchymal cells.
hypovolemia, hyper-reninemia, and metabolic This process gives a 15 branch generation. At
alkalosis) is a group of tubulopathies that 20–22 weeks of gestation, branching morphogen-
involve the regulation and expression of vari- esis is completed and will result in the collecting
ous tubular transporters located in the ascend- system. Mesenchymal cells that are in close con-
ing limb of Henle and the corresponding genes tact with the invading UB undergo an epithelial
(upregulation of NKCC2 and ROMK and transformation. The induced MM gives the neph-
downregulation of Barttin and ClC-Kb). rons through the consecutive stages of condensa-
Bartter syndrome (polyhydramnios and hyper- tion, renal vesicle, vascular cleft, and S-shaped
calciuria) is seen in fetal and neonatal forms. body. The nephrons develop in successive stages
Treatment consists of salt adaptation, water from the inner to the outer area of the kidney in
and potassium intake, and amiloride and/or parallel with the vascular system. The glomerular
indomethacin use. capillary tuft is formed via recruitment and prolif-
• Congenital abnormalities of the kidney and eration of endothelial and mesangial cell precur-
urinary tract (CAKUT) are a range of anoma- sors. In human, the primitive glomerulus appears
lies including malformations of ureteropelvic at approximately 9–10 weeks of gestation.
junction, of vesicoureteric junction, or of Nephrogenesis is completed by 34–36th weeks
vesicourethral area and dysplastic kidneys. of gestation with about 60% of the nephrons
• Renal maldevelopment and exposure to factors being formed during the third trimester of preg-
that can alter fetal and neonatal renal functions nancy (Saxen 1987; Merlet-Benichou et al. 1999).
and structure increase the risks of hypertension Once nephrogenesis is complete, stroma cells dif-
and chronic renal disease in adulthood. Long- ferentiate into fibroblast-, pericyte-, and
term follow-up is thus required and early lymphocyte-like cells. At birth the final nephron
markers of nephron dosing and renal injury varies from 500,000 to 1 million per kidney. Such
variation in nephron number is due to genetic
Pathophysiology of Fetal and Neonatal Kidneys 3

Fig. 1 Kidney
development: simplified Nephron deficit
schematic process of CAKUT
normal and pathologic
kidney development
Abnormal stromal
Abnormal Csyts development
Branching development Nephrogenesis failure (metaplasic tissue)

Genes
Fetal environment
Drugs, toxic
Urinary obstruction

Ureteric bud Metanephric mesenchyme

Branching Nephrogenesis
morphogenesis (epithelial differentiation)

Collecting duct system Nephron Stroma tissue

Kidney and Urinary tract

factors, to the fetal environment, or to a combina-


tion of both (Chevalier 1996). Several genes and Renal Physiology
molecular pathways control the formation of the
renal collecting system and nephrogenesis, such The Fetus
as transcriptional factors (PAX-2, WT1), growth
factors (IGF, EGF, TGF, GDNF, FGF), onco- During intrauterine life, the homeostasis of the
genes, extracellular matrix, and vascular factors fetus is assigned to the placenta. Glomerular fil-
(Table 1) (Burrow 2000). These factors act at a tration rate, renal blood flow, and tubular func-
specific time of kidney development especially tions progress with renal growth and
when the UB interacts with the adjacent nephrogenesis. The kidney is involved in urine
MM. For instance, blockade of VEGF receptor, production which is essential in fetal well-being
inhibition of the renin-angiotensin system (RAS), and in hormonal production (1.25 OH vitD3 and
and knockout mice for COX-2 gene expression erythropoietin). The formation of the urine, the
are associated with impaired nephrogenesis main component of amniotic fluid in the third
including glomerular cysts, dysplastic tubules, trimester, begins by 12 weeks of gestation. The
and tubular dysgenesis (Dinchuk et al. 1995; urinary flow rate increases tenfold from 6 ml/h at
McGrath-Morrow et al. 2006; Pryde et al. 1993). 20 weeks to 60 ml/h at 40 weeks of gestation,
which expresses an optimal structural and func-
tional renal development. In the near-term period,
the fetal kidney shows sufficient glomerular and
4 F. Boubred and U. Simeoni

Table 1 Syndromes and gene defects associated with abnormal kidney development (CAKUT)
Syndrome Gene defects Kidney malformations
Renal-coloboma syndrome PAX 2 Hypoplasia, VUR
Renal cysts and diabetes syndrome HNF1b Dysplasia, hypoplasia
Branchio-oto-renal syndrome EYA1, SIX1 Unilateral or bilateral agenesia/
dysplasia
Renal tubular dysgenesis RAS components Tubular dysplasia/dysgenesis
Campomelic dysplasia SOX9 Dysplasia, hydronephrosis
Townes-Brocks syndrome SALL1 Dysplasia, hypoplasia, VUR
Simpson-Golabi-Behmel syndrome GPC3 Medullary dysplasia
Kallmann syndrome KAL1,FGFR1, PROK Renal agenesis
Fraser syndrome FRAS1 Dysplasia, renal agenesis
Alpert syndrome FGRG2 Hydronephrosis
Alagille syndrome JAGGED1 Cystic dysplasia
Meckel-Gruber syndrome MKS1,MKS3 Cystic renal dysplasia
Hypoparathyroidism, deafness, and renal anomaly GATA3 Dysplasia, VUR
syndrome (HDR)
DiGeorge syndrome 22q11 Agenesia, dysplasia, VUR
Beckwith-Wiedemann syndrome P57 Dysplasia
Pallister-Hall syndrome GLI3 Dysplasia
Nail-patella syndrome LMX1B Agenesia, glomerular anomalies
Smith-Lemli-Opitz syndrome 7 hydroxy-cholesterol Agenesia, dysplasia
reductase
Zellweger syndrome PEX1 Cystic dysplasia, VUR
Glutaric aciduria type II Glutaryl CoA Cystic dysplasia
dehydrogenase
STAR syndrome FAM58A CAKUT
Hypothyroidism PAX8 Renal agenesis
– SOX17, DSTYK, BMP4 CAKUT

tubular development to allow the adaptation to and vasorelaxing factors such the prostaglandins,
extrauterine life (Brophy and Robillard 2004). nitric oxide, and other factors (Fig. 2). Highly
expressed during the active development of the
kidney, these vasoactive factors participate to
Glomerular Function nephrogenesis and in the regulation of intrarenal
vascular blood flow (essential for the develop-
During fetal life, the glomerular filtration rate ment as well).
(GFR) increases progressively, while renal vascu- In the fetal kidney, the RAS is the principal
lar resistances (RVRs) remain elevated. The fetal vasoconstrictive factor; it is upregulated during
GFR is mainly driven by nephrogenesis. Systemic active nephrogenesis and downregulated at the
blood pressure, around 40–60 mmHg, and renal end of this process. The RAS maintains renal
blood flow are low in comparison with the new- blood flow and renal pressure perfusion. The
born or the adult. For instance, in sheep, the fetal fetus is able to release renin into the fetal circula-
kidneys receive 3% of cardiac output compared to tion. Renin does not cross the placenta and the
15% in the neonatal period. Low RBF is associ- fetal plasma renin levels are higher than maternal
ated with elevated RVRs. This state is due to a levels. Angiotensin II increases mainly the glo-
subtle equilibrium between vasoconstrictive fac- merular efferent tone which is highly sensitive to
tors including the renin-angiotensin system angiotensin. Renal sympathetic nervous system
(RAS) and renal sympathetic nervous system increases renal vascular tone, in afferent and
Pathophysiology of Fetal and Neonatal Kidneys 5

Prostaglandins Angiotensin II

aa CHP BSHP ea

Norepinephrine
NO Endotheline
Kinins
ANP
Vasoconstriction aa : Afferent arteriole
ea : Efferent arteriole
CHP: Corpuscular Hydrostatique Pressure
Vasodilatation :Oncotic pressure
BSHP: Bowman Space Hydrostatic Pressure

Fig. 2 Determinants of glomerular filtration with preglomerular and postglomerular vasoactive forces

efferent arteriole. The fetal renal vasculature is and vasodilates the afferent arteriole. In sheep,
more sensitive to alpha2-adrenoreceptor stimula- inhibition of NO increases renal vascular resis-
tion than the neonatal one. Upregulation of tance, impairs renal function, and reduces sodium
alpha2-receptors is associated with down- excretion. Other factors including endothelin and
regulation of beta2-adrenoreceptors. Adenosine atrial natriuretic factors modulate renal hemody-
vasodilates glomerular arterioles in physiologic namic as well.
state but increases tone of afferent glomerular
arterioles in stress conditions, responsible for
reduced GFR. Tubular Function
Vasoconstrictive forces are counterbalanced by
vasodilating factors that principally act on the The tubular system has different secretion and
glomerular afferent arteriole to maintain a suffi- reabsorption activities depending on the tubular
cient renal blood flow. Prostaglandins, nitric oxide segment. For example, the proximal tubule is
(NO), and kallikrein-kinin system are principal principally involved in regulation of fluid, elec-
factors. Prostaglandins, in particular E2 and I2, trolytes, amino acids, glucose, and the distal
are of major importance. These prostaglandins are tubules and collecting system in fluid and tonicity
produced by the placenta, membranes, and the regulation. Tubular transporters mature differ-
fetus, from the action of two enzyme isoforms, ently during the development of the nephrons.
the cyclooxygenase types 1 and 2 (COX-1 and Distal tubular phosphorus and potassium channels
COX-2), and their production is stimulated by mature early to make them available for cellular
angiotensin II (34). COX-2 is constitutive in functions. These features are responsible in part
fetal kidney and appears essential for the devel- for higher fetal plasma potassium and phosphorus
opment and function of the kidney (Khan et al. levels than in the mother. Renal tubules are the
2001). Administration of COX inhibitors during target of hormones including RAS, aldosterone,
pregnancy decreases renal blood flow, impairs prostaglandins, atrial natriuretic peptide, and cor-
renal function, and induces oligohydramnios. tisol (maturational effect). The sensitivity of renal
Nitric oxide is synthetized by endothelial cells tubules to such hormones matures progressively
6 F. Boubred and U. Simeoni

along gestation. Finally the relative tubular imma- However, the major role of the kidney in fetal
turity associated with hypotonic urines calcium homeostasis is the production of 1.25
(100–250 mOsm/kg H2O) allows the production (OH)2D3 rather than renal regulation of calcium
of amniotic fluid at a sufficient rate which is excretion.
essential for fetal well-being (Brophy and During fetal development, the placenta regu-
Robillard 2004). lates the fetal acid-base balance. The reabsorption
The excretion of sodium is high during fetal of bicarbonates and chloride in the proximal
life but decreases with renal development. This tubule increases with gestational age, which
state is related to various factors including high allows the fetus to participate to acid-base homeo-
circulating sodium concentrations and high sensi- stasis, near term. This function is mainly related to
tivity to natriuretic factors, large extracellular the maturation of the carbonic anhydrase activity.
fluid volume, relative insensitivity to hormonal The fetus produced hypotonic urine at an ele-
factors, and dysmaturity of tubular sodium vated rate of 1.5 l every day. The urine concentra-
reabsorption. In opposition with adult physiology, tion capacity is blunted in the immature kidney.
the sodium is mainly reabsorbed in the distal This defect is related to various factors including a
portion of the immature tubules. The sodium/ low sensitivity of the collecting duct to arginine
hydrogen exchangers (NHE, four different iso- vasopressin (AVP), a structural immaturity of the
forms) play an important role. These exchangers loop of Henle with preferential distribution of
mediate the exchange of one sodium for one blood flow to the inner cortex, a low gradient
hydrogen and contribute to the acidification of concentration in the medulla due to limited pro-
urine. The NHE three channel, located at the api- tein intake to generate significant amounts of urea,
cal or luminal membrane of tubular cells, is and a low expression of aquaporin 2 (AQ2). AQ2,
thought to be responsible for the bulk of tubular a water channel, is located in the apical membrane
reabsorption of sodium. It is downregulated in the of collecting duct cells and is involved in water
proximal portion of the immature tubules when it reabsorption. Humans who lack aquaporins
is markedly upregulated after birth. Its expression (AQs) have a urinary concentrating defect.
and activity are dependent on tubular Na+, K+- Expression of AQs is regulated by the AVP via
ATPase which is downregulated in immature kid- the V2 receptor. AVP is highly synthesized during
ney (Hoster 2000). the last trimester in pregnancy (Nielsen and
The balance of potassium is positive in the Frokaier 2002). The immaturity of tubular func-
fetus, due to active transplacental transfer and tions and its maturation explain the high rate of
renal immaturity. They both make potassium amniotic fluid production which decreases at the
available for cell growth and cell functions. end of gestation.
Renal secretion is low and tends to increase
toward term. Potassium excretion increases with
glomerular and tubular surface area, with matura- The Newborn
tion of tubular Na+, K+-ATPase activity, and with
the progressive tubular sensitivity to aldosterone. Glomerular Function
As for potassium, the fetus is characterized by After birth hemodynamic changes (increased
a positive phosphorus balance resulting from a blood pressure and renal blood flow) and struc-
transplacental transport against a concentration tural changes contribute to postnatal maturation of
gradient, a high rate of tubular reabsorption via a GFR. As nephrogenesis is terminated at birth,
sodium-phosphorus co-transporter, and a relative postnatal structural change is based on the matu-
parathyroid insufficiency. Calcium is important ration of preexisting renal structures. This process
for the adequate mineralization, growth, and includes increase in glomerular membrane perme-
fetal skeleton. Vitamin D-dependent calcium- ability, in corpuscular glomerular diameter (espe-
binding proteins, involved in transepithelial cal- cially of glomeruli from the outer area of cortex),
cium transfer, are present in fetal kidney. in filtration surface area, and in intrarenal
Pathophysiology of Fetal and Neonatal Kidneys 7

redistribution of blood flow. Glomerular size beta trace protein are not used routinely in neo-
reaches adult values at the age of 3 years. These nates. Clearance of inulin is the gold standard to
structural changes result in enhanced filtration evaluate GFR. Inulin is inert, being not metabo-
surface area. lized and neither reabsorbed nor secreted by the
Transition of fetal circulation to extrauterine renal tubules; however, it has limited applications
life is characterized by a rapid increase in systemic in clinical practice. Cystatin C is produced by all
blood pressure and renal blood flow. The rapid nucleated cells and is independent of muscle
change in RBF is associated with postnatal mass. It is freely filtered and reabsorbed but catab-
decrease in renal vascular resistances (decrease olized in the kidney, which prevents clearance
in glomerular vasoconstriction). Glomerular cap- measurement. Serum levels are high during the
illary hydrostatic pressures increase as well but neonatal period and stabilize at 12 months of age.
remain lower than in the adult (Solbaug and Jose Cystatin C and beta trace protein are being evalu-
2004). ated in newborn and preterm infants. Current data
After birth GFR rapidly increases (2.5- to consider cystatin C as a valuable tool to assess
threefold increase) from 20 ml/min/1.73 m2 to GFR in newborn and preterm infants (Filler et al.
half of adult values at the end of the neonatal 2016).
period (Guignard 1975). In preterm infants GFR
correlates with gestational and is at birth less than Tubular Function
15 ml/min/1.73 m2. Postnatal maturation depends The postnatal maturation of renal tubules follows
on gestational age with a delayed maturation the maturation of GFR. Postnatal maturation is
being possibly observed in very low birth weight characterized by a tenfold increase in proximal
infants (Gubhaju et al. 2014). In term infants, tubular length and diameter within the weaning
plasma creatinine concentrations are elevated at period.
birth (60–70 mcmol/l) and stabilize at the end of After birth, the regulation of homeostasis is
the first week (30–40 mcmol/l). In preterm transferred from the placenta to the kidney. Post-
infants, creatinine levels are elevated on the first natal fluid and electrolytic adaptation is character-
day (90–110 mcmol/l), increasing transiently ized by a transient increase in diuresis during the
within days 3–5 (reaching 130–150 mcmol/l) first week of life due to the contraction of the
and then decrease progressively (Gallini et al. extracellular volume. Commonly neonates lose
2000). Neonates and especially preterm infants around 5–10% of their birth weight. Very low
are in a state of physiologically renal insuffi- birth weight may undergo a higher weight loss
ciency. A delicate balance of vasoconstrictor and of up to 15% of their birth weight. Fractional
vasodilator forces which maintains GFR and excretion of sodium in neonates is around 1%
immature renal structure makes newborn and and decreases progressively in relation to a rapid
especially preterm infants at high risk to develop maturation of proximal tubules. Fetal exposure to
renal dysfunction in specific conditions (hypoten- glucocorticoids at the end of gestation accelerates
sion, hypovolemia, perinatal asphyxia, and neph- the maturation of tubular transporters (NHE, Na+,
rotoxic drugs). K+-ATPase, etc.) (Brophy and Robillard 2004).
Serum creatinine is widely used to assess glo- The neonate is able to achieve maximal dilu-
merular filtration rate (GFR) in neonates. Clear- tion with urine osmolarity as low as 40–60 mOsm/
ance of endogenous creatinine can be used in l. Diluting capacity tends to mature rapidly in the
clinical practice and needs strict 12–24-h urine newborn and in the preterm infant who at 36th
collection which can be difficult in preterm postconceptional age have the same dilution
infants. Creatinine is a metabolite of creatine capacity than adult. Within the limits allowed by
located in skeletal muscle and is filtered and of GFR, neonates, even preterm infants, tolerate a
secreted in part by renal tubular cells. Muscle large range of fluid intakes (150 ml/kg/day) with-
mass influences serum creatinine levels. Other out major alterations in water and electrolyte
markers of GFR including inulin, cystatin C, and parameters. Diluting capacity is greater than
8 F. Boubred and U. Simeoni

Fig. 3 Renal mechanism Nephrotoxic Maternal gestational


of systemic hypertension Maternal
nutritional status drugs diabetes
and renal damage, the
“Brenner hypothesis” Congenital renal
IUGR Reduced nephron number anomalies
Preterm birth ?

Filtration surface area

Single nephron
Arterial
glomerular filtration rate
blood pressure
(SNGFR)

Glomerular hypertension

Proteinuria
Glomerulosclerosis

concentrating capacity, which is limited to


400–600 mOsm/l. Children reach the adult maxi- Pathophysiology
mal urine concentration ability
(1300–1400 mOsm/kg) by 2 years of age. This The Immature Kidney
characteristic may be explained by reduced tonic-
ity of the medullary interstitium, low expression In the preterm infants, nephrogenesis is incom-
of water channels (aquaporins), and relative tubu- plete at birth and has to continue in an unfit envi-
lar insensitivity to ADH. Higher production of ronment. Nephrogenesis may be altered in
prostaglandin E2 in neonate may inhibit the tubu- preterm infants (Rodriguez et al. 2004; Sutherland
lar effect of ADH. Low neonatal concentrating et al. 2011). Experimental studies show discordant
capacity is of limited importance in healthy neo- findings (Stelloh et al. 2012; Gubhaju et al. 2009).
nates. However, neonates and especially preterm In rodents only one study has shown reduced
infants are more vulnerable to insufficient water nephron endowment in pups born preterm
intakes and extrarenal water loss (skin losses, (Stelloh et al. 2012). Such a finding has not been
diarrhea, etc.) with the risk of hypernatremic found in preterm baboon (Gubhaju et al. 2009).
dehydration and osmotic diuresis and favored by The degree of immaturity may explain such dis-
a lower threshold for renal glucose excretion. cordant findings. In contrast with the baboon
The newborn has a diminished threshold for model, preterm birth occurred during the early
bicarbonates. The large extracellular fluid (ECF) stage of nephrogenesis in the rodent models.
volume expansion may result in depressed proxi- These findings suggest that extremely preterm
mal tubular bicarbonate reabsorption. When ECF infants are at high risk of reduced nephron endow-
is contracted, renal reabsorption increases and ment. Data are limited in preterm infants; autopsy
urinary pH becomes more acidic. In preterm findings have shown an alteration of glomerular
infants, immaturity of carbonic anhydrase may structure and acceleration of nephron maturation
prolong the depressed tubular bicarbonate with a possibly reduced nephron endowment
reabsorption. The normal range for plasma bicar- (Rodriguez et al. 2004; Sutherland et al. 2011).
bonate concentration is lower in preterm infants Impairment of mesenchymal cell differentiation
(16–20 mmol/l) than term infants (21–24 mmol/l). has been suggested. Moreover it is not excluded
Pathophysiology of Fetal and Neonatal Kidneys 9

Cinulin
60
Term

40 Premature

70 20

60 Days
C inulin , ml/min per 1.73m2

5 10 15 20
50

40

30

20

10

2 4 6 8 10 12 14 16 18 20 22 24 26
Postnatal age, days

Fig. 4 Postnatal glomerular filtration rate in neonates (C inulin: inulin clearance) (From Guignard et al. 1981)

that environment factors to which preterm infants inhibitors (indomethacin) for closure of a patent
face up during the neonatal period including ductus arteriosus impairs renal function, leads to
infection/inflammation, oxidative stress, nephro- oliguria, and reduces RBF and GFR. Ibuprofen,
toxic drug use, or undernutrition can influence another COX inhibitor, has moderate renal
nephrogenesis and thus nephron endowment. For effects. However, these adverse renal effects can
instance, neonatal undernutrition has been shown be prolonged up to 1 month and can limit renal
to be associated with reduced kidney volume excretion of some medications (Giniger et al.
(a surrogate of nephron mass) in infants born 2007; Vieux et al. 2011). In animal, neonatal
preterm (Bacchetta et al. 2009). Collectively the indomethacin administration results in reduced
current findings suggest that the extremely pre- nephron number in adult rats unlike ibuprofen
term infants and preterm infants exposed to (Kent et al. 2014).
adverse neonatal complications have a high risk Postnatal hydro-electrolytic adaptation is char-
of nephron deficit. acterized by an excessive water and sodium losses
In comparison with term neonates, GFR is during the first 10 days after birth. Preterm infants
lower in preterm infants and increases progres- are prone to water and salt wasting with high risk
sively after birth (Gubhaju et al. 2014; Bueva of early hypernatremia and late hyponatremia.
and Guignard 1994) (Figs. 4, 5 and 6). Structural Fractional excretion of sodium is elevated in pre-
maturation drives the progressive maturation of term infants (5% vs <1% in term infants) and
GFR in very preterm infants in whom postnatal reaches term values progressively after days
nephrogenesis continues after birth. Blood pres- 15–21. A postnatal contraction of the extracellular
sure and renal blood flow are low in preterm fluid volume occurs during the first postnatal days
infants. The RVRs are high and the RAS and translates into a 2–3% daily weight loss. The
upregulated. The immature kidney is particularly optimal maximum weight loss is unknown; a
dependent on vasodilator forces to maintain a maximum limit of 10–15% total weight loss is
sufficient GFR. Administration of prostaglandin common especially in extremely preterm infants
10 F. Boubred and U. Simeoni

70

1000-1500g

Creatinine clearance, ml/min × 1.73m2


60 1501-2000g
2001-2500g

50 term

40

30

20

10

0
0 1-2 8-9 15-16
Age, days

Fig. 5 Postnatal maturation of glomerular filtration rate in term and preterm infants (Bueva and Guignard 1994)

and should be not exceeded. Fluid and electrolyte et al. 2015; Sweet D and the working group on
balance has to be thus closely followed up and prematurity 2007). Glomerular and tubular conse-
adapted. Moreover, the kidney of preterm infants quences of immaturity are shown in Table 2.
cannot efficiently excrete excessive water and Preterm infants are more likely to receive drugs
sodium loads which increase the risk of in neonatal intensive care unit. Most drugs are
bronchopulmonary dysplasia, intracerebral hem- eliminated by the kidney and undergo glomerular
orrhage, and necrotizing enterocolitis. It is com- filtration or tubular metabolism both being imma-
mon to introduce sodium, potassium, and ture in preterm infants. Circulating levels of drugs
phosphorus by postnatal day 2 and to adapt indi- such as vancomycin or aminoglycosides need to
vidually electrolyte intake (especially sodium). be monitored to optimize dosage and to reduce
However, nutritional practices have recently toxicity.
changed with early introduction of high protein Glomerular and tubular consequences of pre-
and caloric intakes, which stimulates anabolism maturity are blunted by maternal administration of
and prevents early catabolism. This new nutri- synthetic glucocorticoids (GCs) (betamethasone
tional strategy enhances cellular use of potassium and dexamethasone). Prenatal administration of
and phosphorus and increases the risk of early GC improves systemic blood pressure, RBF, and
hypokalemia and hypophosphatemia especially GFR and accelerates the maturation of tubular
in intrauterine growth restricted preterm infants functions with enhanced proximal reabsorption
(Boubred et al. 2015). Potassium and phosphorus of sodium and excretion of potassium. This treat-
intake can be thus introduced early from birth ment limits dysnatremia and hyperkalemia events
onwards for some infants. Rapid growth induces in preterm infants. Increased expression and activ-
sodium cell utilization and together with sodium ity of sodium exchanger, Na+, K+-ATPase activ-
wasting favors the occurrence of late hypo- ity, and renal sympathetic nerve activity are
natremia. Fluid, minerals, and electrolyte balances involved (Catarelli et al. (2002).
should be adapted tightly and individualized to
promote optimal growth and deficiency (Boubred
Pathophysiology of Fetal and Neonatal Kidneys 11

150,0
140,0
GA <27 wks
Serum Creatinine (mmol/l) 130,0
GA 27-28 wks
120,0 GA 29-30 wks
110,0 GA 31-32 wks
100,0
90,0
80,0
70,0
60,0
50,0
40,0
30,0
0 1 2 3 4 5 6 7 10 17 24 31 38 45 52
Days of life
45,0
GA <27 wks
Creatinine Clearance (ml/min/1.73 m2)

40,0
GA 27-28 wks
35,0 GA 29-30 wks
30,0 GA 31-32 wks

25,0

20,0

15,0

10,0

5,0

0,0
3 7 10 17 24 31 38 45 52
Days of life

Fig. 6 Postnatal maturation of glomerular filtration rate in extremely low gestational age neonates (Gallini F et al.
Pediatre Nephrol 15:119–124)

Congenital Tubulopathies consists of salt adaptation, water and potassium


intake, and amiloride and/or indomethacin use.
Congenital tubular diseases are rare and include Primary pseudo-hypoaldosteronism is an auto-
various diseases according to defect in epithelial somal recessive disease due to defective epithelial
tubular channels (Rodriguez-Soriano 2000). ENaC channel (in collecting ducts). This syn-
Bartter syndrome is a group of tubulopathies drome is characterized by life-threatening salt
involving the regulation and expression of various wasting, risk of dehydration and arterial hypoten-
tubular transporters located in the ascending limb sion, severe hyperkalemia, distal tubular acidosis,
of Henle and the corresponding gene and hyperaldosteronism. This disease is treated by
(upregulation of NKCC2 and ROMK and down- sodium supplementation. When ENaC activity is
regulation of Barttin and ClC-Kb). Bartter syn- increased, sodium reabsorption is exaggerated
drome is characterized by salt wasting, by with hypertension and metabolic hypokalemia.
hypovolemia with a low or conserved blood pres- Renin activity and aldosterone levels are
sure, by hyper-reninism, and by metabolic alkalo- decreased. Liddle syndrome, an autosomal domi-
sis. Polyhydramnios and hypercalciuria are often nant disease, displays such features. Treatment
observed in fetal and neonatal forms. Treatment consists in salt restriction and amiloride.
12 F. Boubred and U. Simeoni

Table 2 Glomerular and tubular consequences of immature kidney


Fluid-electrolytes therapy in Glomerular immaturity:
preterm infantsa consequences Tubular immaturity: consequences
D1–D2: Reduced ability to salt/water Sodium wasting
Volume: 80–100 ml/kg/d overload excretion
Na: 0 Hyperkalemia Metabolic acidosis
K: 0 (except if Reduced urine concentration capacity with
<3.5 mmol/l) conservative urine dilution capacity
D3–D5: High sensitivity to vasoactive Glycosuria
Volume: 120–140 ml/kg/d drugs
Na: 2–4 mmol/kg/d Increased risk of renal failure Elevated urinary calcium excretion
K: 1–2 mmol/kg/d
D5: Reduced clearance of drugs High sensitivity to diuretics
Volume: 140–170 ml/kg/d
Na: 4–6 mmol/kg/d
K: 1–3 mmol/kg/d
a
Fluid, micronutrient, and electrolyte intakes are individually adapted

Nephrogenic diabetes insipidus, an X-linked or such a situation. In human, congenital


autosomal transmitted disease, is characterized by ureteropelvic junction obstruction is associated
polyuria-polydipsia, dehydration, irritability, and with renal histologic changes including decreased
failure to thrive. These symptoms are due to a glomerular density, interstitial fibrosis, and tubu-
defect in the arginine vasopressin-V2 receptor or lar dilatation (Peters et al. 1992).
in AQ2. A genetic and developmental mechanism has
been proposed for CAKUT. Various defects in
specific genes involved in renal development
Parenchymal Consequences of Urinary affect kidney parenchyma and urinary tract devel-
Tract Anomalies opment (few examples in Table 1). However, the
pathogenesis of this syndrome is multifactorial
Congenital abnormalities of the kidney and uri- and involves single nucleotide variants and epige-
nary tract (CAKUT) are a wide range of anoma- netic mechanism through which environment can
lies including malformations of ureteropelvic impair the development of the kidney and urinary
junction, of vesicoureteric junction, or of tract (Nicolaou et al. 2015). The quality of the
vesicourethral area and dysplastic kidneys. ureteric bud-mesenchymal interaction, the posi-
tion at which arises the UB from the pronephros,
Renal Structure Changes and the number of branching morphogenesis are
The pathophysiological mechanism of CAKUT critical for the kidney development. Failure of
remains unknown. One can postulate that urinary induced ureteric bud outgrowth leads to renal
obstruction affects the interaction UB-MM with agenesis. An aberrant outgrowth of more than
failure in the induction of metanephrogenic blas- one UB may result in double collecting system
tema. After an early compliance stage, obstruction and duplication of the ureter. An ectopic position-
of the collecting system induces urinary hyper- ing of the UB is associated with renal tissue mal-
pressure and muscular hypertrophy. In fetal formation, abnormalities of the ureterovesical
lamb, early and complete ureteral obstruction junction, and a nephron deficit related to reduced
impairs renal architecture with abnormal tubular branching morphogenesis (Fig. 1).
differentiation, inflammation, and reduced neph-
ron endowment. Changes in renal expression of Renal Function Changes
transcription factors (Pax2, Wnt) involved in early The outcome of CAKUT depends on the impor-
stage of nephrogenesis have been demonstrated in tance of urinary obstruction, on the kidney
Pathophysiology of Fetal and Neonatal Kidneys 13

structure, on fetal renal functions neonatal death. Selective COX-2 inhibitors and
(oligohydramnios, urinary sodium concentra- nonselective nonsteroidal anti-inflammatory
tions, fetal beta2-microglobulin), and on defects drugs (NSAIDs), ACE-Is, and AT1R antagonists
in other organs. Early occurrence of urinary tract are the main drugs affecting the development and
obstruction, bilateral renal dysplasia, reduction of the function of perinatal kidney [review 32].
the kidney size, and oligohydramnios (a surrogate Nonsteroidal anti-inflammatory drugs
of severe fetal renal insufficiency) is associated (NSAIDs) can be used as antalgic or as tocolytic
with poor renal prognosis. Infants with severe agent in pregnant woman (indomethacin). The
pathology often die in the perinatal period as a renal side effects of in utero exposure to NSAIDs
consequence of Potter’s sequence (pulmonary vary from transient fetal oligohydramnios to
hypoplasia resulting from oligohydramnios and severe and lethal renal failure in babies. Quickly
fetal renal function insufficiency). Survivors may after the initiation of indomethacin therapy, a sig-
develop renal failure during childhood. Dialysis nificant reduction of fetal urine production occurs.
during neonatal period should be discussed with This effect, frequent and transient in many cases,
parents and the multidisciplinary nephrology is the basis of treatment of polyhydramnios. The
team before and after birth. available information originates principally from
Infants with severe obstructive nephropathy retrospective studies and indicates a rate of inci-
are prone to acid-base and hydro-electrolytic bal- dence from 1.5% to 20%. Experimental and
ance abnormalities and to impaired GFR. Com- human studies show that NSAIDs decrease fetal
plete and prolonged fetal ureteral obstruction is GFR (consequence of renal blood flow reduction)
associated with decrease RBF and GFR propor- and increase urine osmolality (enhanced activity
tional to loss of parenchyma. The RAS is of arginine vasopressin). The risk of neonatal
upregulated in such situations. Renal dysfunc- renal failure seems increasing with prolonged
tions lead to type IV renal tubular acidosis (hyper- and cumulate doses, short time period between
chloremic metabolic acidosis) with hyperkalemia, treatment and delivery, preexisting fetal distress,
salt wasting, and defect in urine concentration and low birth weight. In addition to the changes of
responsible for water wasting and nephrogenic renal functions, a singular renal nephrotoxicity
diabetes insipidus. Various changes in tubular dysplasia attributable to NSAIDs has been
channel exchanger, decreased NA+, K+-ATPase described: various degrees of ischemic injuries
activity, unresponsiveness of the damaged distal and fibrosis of medullary area, cortical necrosis,
nephron to aldosterone with secondary hyper- focal tubular and glomerular microcysts of devel-
aldosteronisme, loss of H+ ATPase on the surface oping nephron, loss of differentiation between
of intercalated cells, and downregulation of proximal and distal tubules, and decreased renal
aquaporins have been described. Treating and mass. These lesions are associated with increased
preventing renal infection, adapting protein expression of renin in the juxtaglomerular appa-
intake, and limiting the use of or close monitoring ratus. The severity of renal hypoperfusion induced
of nephrotoxic drugs aim to preserve renal by NSAIDs may lead to irreversible glomerular
functions. and tubular injuries. Such effects have been noted
recently after prenatal exposure to the selective
inhibitors of COX-2, new generations of NSAIDs,
Pharmacological Interactions proposed as an alternative to nonspecific NSAIDs
with the aim of preventing adverse side effects.
The number of pregnant women and women of When administered during pregnancy, inhibi-
childbearing age receiving drugs is constantly tors of the RAS (ACE-Is and AT1R antagonists)
increasing. These drugs can cross the placenta can induce a variety of serious fetal complica-
and can impair potentially the function and struc- tions, with a high degree of mortality rate. These
ture of the fetal kidney. The newborn may in turn complications named angiotensin-converting
express renal failure with various severities up to enzyme inhibitor fetopathy include intrauterine
14 F. Boubred and U. Simeoni

growth retardation, profound fetal and neonatal of follow-up was 7 years) is insufficient to con-
hypotension refractory to any treatment, renal clude definitely on the side effects at adulthood.
failure, oligohydramnios with limb deformities Antenatal maternal administration of ampicil-
and pulmonary hypoplasia, hypocalvaria, and lin and aminoglycosides reduces nephron number
increased rate fetal loss. Renal histology shows (20–30%) and induces hypertension in adult rat
typically proximal tubular dysgenesis. Numerous offspring. Reduced nephron number results from
cases have been reported with a wide range of a defect in UB branching morphogenesis affecting
adverse fetal and neonatal renal effects varying the first branching division. Administration dur-
from transient oligohydramnios (when treatment ing the late stage of nephrogenesis does not
was arrested rapidly) to severe (requiring perito- induce a nephron deficit. Other aminoglycosides,
neal dialysis) and lethal renal failure. The magni- such amikacin and netilmicin, have less adverse
tude of these adverse renal effects cannot be renal effects. Antenatal maternal administration of
quantifiable precisely, but the risk increases by ampicillin is associated with 20% reduction of
chronic administration and the stage of fetal expo- nephron number in rat offspring; apoptosis mech-
sure, especially during the second and third tri- anism of MM is the suspected. Such experimental
mester. In contrast, elective exposure in the first observations have not been found in human. Other
trimester of gestation is not associated with an drugs, such as chlorambucil and antineoplastic
enhanced risk of teratogenicity. When therapy is drugs, can impair renal development with urinary
interrupted rapidly before 2nd trimester, poor or tract malformation and renal agenesis.
no adverse renal effects are noted. In summary, in most cases, newborn do not
Experimental studies have shown that maternal express clinical renal failure postnatally. How-
antenatal administration of various drugs can ever, experimental studies suggest that prenatal
affect nephrogenesis in the absence of renal dys- exposure to certain drugs may decrease nephron
functions in the fetus and newborn infants. In endowment. One must be careful in extrapolating
rodents and sheep, antenatal maternal administra- data from animal to human, since major interspe-
tion of glucocorticoids (GCs) at a specific time of cies differences in renal sensitivity to drugs exist.
pregnancy reduces nephron number and induces However, long-term follow-up of infants exposed
hypertension in adult offspring. The mechanism in utero to drugs is needed.
by which GCs alter the fetal kidney structure is
unclear. Several pathways are suspected. In utero
exposure to GC may (1) disturb cell differentia- Long-Term Consequences of Nephron
tion/proliferation ratio at the expense of reduced Number Reduction
proliferation, (2) reduce ureteric bud branching,
(3) alter gene expression of specific genes It is suspected for a long date, since the works of
involved in kidney development (GNDF), Brenner et al. in the 1980s, that a reduced nephron
(4) decrease renal AT1R and renin gene expres- number increases the risk of hypertension and
sion, or (5) alter epigenetic marks. chronic renal insufficiency in adulthood (Brenner
Maternal administration of cyclosporine A et al. 1988). Reduced nephron number is associ-
(CsA) during gestation (E14–18, and E20–24) ated with renal changes including single nephron
affects fetal growth and nephrogenesis leading to glomerular filtration hyperfiltration (SNGHF) and
a permanent nephron deficit in rat offspring (aver- glomerular and tubular hypertrophy. Such hemo-
age of 25–30%). CsA may block epithelial trans- dynamic changes are responsible for glomerular
formation of the metanephric mesenchyme. In injury through increase in glomerular capillary
human, outcome of infants exposed in utero to hypertension. Over a long time, a vicious cycle
CsA has not shown any changes of renal function takes place leading to glomerular sclerosis,
and morphology in childhood. But data are too chronic renal insufficiency, and hypertension
scarce, and long-term follow-up (the maximal age (cf Fig. 3). In human, an inverse relationship has
been established between nephron number and
Pathophysiology of Fetal and Neonatal Kidneys 15

hypertension (Keller et al. 2003). However, this References


relationship is complex and involves various fac-
tors such as nephron dosing and postnatal factors. Bacchetta J, Harambat J, Dubourg L et al (2009) Both
extrauterine and intrauterine growth restriction impair
For instance, congenital agenesis, a situation of
renal function in children born very preterm. Kidney Int
severe nephron deficit, is associated with early 76(4):445–452
development of systemic hypertension and early Boubred F, Vendemia M, Garcia-Meric P et al (2006)
chronic renal insufficiency during infancy of ado- Effects of maternally administered drugs on the fetal
and neonatal kidney. Drug Saf 29:397–419
lescent. It is postulated that a moderate nephron
Boubred F, Saint-Faust M, Buffat C et al (2013) Develop-
deficit at birth is insufficient to induce long-term mental origins of chronic renal disease: an integrative
cardiovascular and chronic renal diseases but con- hypothesis. Int J Nephrol 2013:346067. https://doi.org/
stitutes a factor of vulnerability when other post- 10.1155/2013/346067
Boubred F, Herlenius E, Bartocci M et al (2015) Extremely
natal factors including early catch-up growth or
preterm infants who are small for gestational age have a
overfeeding accelerate the development of these high risk of early hypophosphatemia and hypokalemia.
diseases (Boubred et al. 2013). Acta Paediatr 104:1077–1083
At birth the final nephron number varies from Brenner BM, Garcia DL, Anderson S (1988) Glomeruli
and blood pressure. Less of one, more the other. Am J
500,000 to 1 million per kidney. Such variation in
Hypertens 1:335–347
nephron number is due to genetic factors, to the Brophy PD, Robillard JE (2004) Functional development
fetal environment, or to a combination of both. of the kidney in utero. In: Polin A, Fox W (eds) Fetal
Various perinatal factors have been shown to and neonatal physiology, 3rd edn. W.B. Saunders Com-
pany, Philadelphia, pp 1229–1239
induce nephron deficit including intrauterine
Bueva A, Guignard JP (1994) Renal function in preterm
growth restriction (IUGR), maternal nutrient defi- neonates. Pediatr Res 36:572–577
ciency (vitamin A, iron depletion, zinc defi- Burrow CR (2000) Regulatory molecules in kidney devel-
ciency), maternal protein or global nutrition opment. Pediatr Nephrol 14:240–253
Catarelli D, Chirico G, Simoni U (2002) Renal effects of
deficit, exposure to drugs (see above) or toxic
antenally and postnatally administered steroids. Pediatr
(alcohol), maternal and fetal stress, and maternal Med Chir 24:157–162
gestational diabetes (Merlet-Benichou et al. Chevalier RL (1996) Developmental renal physiology of the
1999). Preterm birth with postnatal adverse envi- low birth weight preterm newborn. J Urol 156:714–719
Dinchuk JE, Car BD, Focht RJ et al (1995) Renal abnor-
ronment (postnatal denutrition, stress, exposure to
malities and an altered inflammatory response in mice
nephrotoxic drugs) and renal and urinary tract lacking cyclooxygenase 2. Nature 378:406–409
malformations are associated with impaired Filler G, Guerrero-Kanan R, Alvarez-Elías AC (2016)
nephrogenesis which may lead to reduced neph- Assessment of glomerular filtration rate in the neonate:
is creatinine the best tool? Curr Opin Pediatr 28
ron endowment.
(2):173–179
Such perinatal events are less likely to affect Gallini F, Maggio L, Romagnoli C et al (2000) Progression
renal functions during the neonatal period but of renal function in preterm neonates with gestational
increase the susceptibility of hypertension and age 32 weeks. Pediatr Nephrol 15:119–112
Giniger RP, Buffat C, Millet V et al (2007) Renal effects of
chronic renal disease in adulthood. Long-term
ibuprofen for the treatment of patent ductus arteriosus
follow-up is needed, with close assessment of in premature infants. J Matern Fetal Neonatal Med
blood pressure and renal parameters (creatinine, 20:275–283
proteinuria) to prevent such adverse evolutive car- Gubhaju L, Sutherland MR, Yoder BA et al (2009) Is
nephrogenesis affected by preterm birth? Studies in a
diovascular and renal diseases. Future studies may
non-human primate model. Am J Physiol Renal Physiol
focus on the development of markers of nephron 297:F1668–F1677
number endowment and of early biomarkers of Gubhaju L, Sutherland MR, Horne RS, Medhurst A, Kent
SNGHF and renal injury with the aim to implement AL, Ramsden A, Moore L, Singh G, Hoy WE, Black
MJ (2014) Assessment of renal functional maturation
preventive strategies).
and injury in preterm neonates during the first month of
life. Am J Physiol Renal Physiol 307(2):F149–F158
Guignard JP (1975) Glomerular filtration rate in the first
three weeks of life. J Pediatr 87:268–272
16 F. Boubred and U. Simeoni

Guignard JP, Gruskin AB, Norman ME (eds) (1981) Pedi- Pryde PG, Sedman AB, Nugent CE et al (1993)
atric nephrology. Martinus Nijhoff, The Hague Angiotensin-converting enzyme inhibitor fetopathy. J
Hoster M (2000) Embryonic epithelial membranes trans- Am Soc Nephrol 3:1575–1582
porters. Am J Phys 279:F74–F52 Rodriguez MM, Gomez AH, Abitbol CL (2004) Histomor-
Keller G, Zimmer G, Gerhard M et al (2003) Nephron phometric analysis of postnatal glomerulogenesis on
number in patients with primary hypertension. N Engl extremely preterm infants. Pediatr Dev Pathol 7:17–25
J Med 348:101–108 Rodriguez-Soriano J (2000) New insight into the patho-
Kent AL, Koina ME, Gubhaju L et al (2014) Indomethacin genesis of renal tubular acidosis-from functional to
administered early in the postnatal period results in molecular studies. Pediatr Nephrol 14:1121–1136
reduced glomerular number in the adult rat. Am J Saxen L (1987) Organogenesis of the kidney. Cambridge
Physiol Renal Physiol 307:F1105–F1110 University Press, Cambridge
Khan KNM, Stanfield KM, Dannenberg A et al (2001) Solbaug MJ, Jose PA (2004) Postnatal maturation of renal
Cyclooxygenase-2 expression in the developing blood flow. In: Polin A, Fox W (eds) Fetal and neonatal
human kidney. Pediatr Dev Pathol 4:461–466 physiology, 3rd edn. W.B. Saunders Company, Phila-
McGrath-Morrow S, Choc C, Molls R et al (2006) VEGF delphia, pp 1243–1249
receptor 2 blockade leads to renal cyst formation in Stelloh C, Allen KP, Mattson DL et al (2012) Prematurity
mice. Kidney Int 69:1741–1748 in mice leads to reduction in nephron number, hyper-
Merlet-Benichou C, Gilbert T, Vilar J et al (1999) Nephron tension, and proteinuria. Transl Res 159:80–89
number: variability is the rule. Causes and conse- Sutherland MR, Gubhaju L, Moore L, Kent AL, Dahlstrom
quences. Lab Investig 79:515–526 JE, Horne RS, Hoy WE, Bertram JF, Black MJ (2011)
Nicolaou N, Renkema KY, Bongers EM et al (2015) Accelerated maturation and abnormal morphology in
Genetic, environmental, and epigenetic factors the preterm neonatal kidney. J Am Soc Nephrol 22
involved in CAKUT. Nat Rev Nephrol 11:720–731 (7):1365–1374
Nielsen S, Frokaier J, Marples D et al (2002) Aquaporins in Sweet D, the working group on prematurity (2007)
the kidney: from molecule to medicine. Physiol Rev European consensus guidelines on the management of
82:205–244 neonatal respiratory distress syndrome. J Perinat Med
Peters CA, Carr MC, Lais A et al (1992) The response of 35:175–186
the fetal kidney to obstruction. J Urol 148:503–509 Vieux R, Fresson J, Guillemin F et al (2011) Perinatal drug
exposure and renal function in very preterm infants.
Arch Dis Child Fetal Neonatal Ed 96:F290–F295

You might also like